• 검색 결과가 없습니다.

Rheumatiod Arthritis: An Updated Overview of Latest Therapy and Drug Delivery

N/A
N/A
Protected

Academic year: 2022

Share "Rheumatiod Arthritis: An Updated Overview of Latest Therapy and Drug Delivery"

Copied!
15
0
0

로드 중.... (전체 텍스트 보기)

전체 글

(1)

Rheumatiod Arthritis:

An Updated Overview of Latest Therapy and Drug Delivery

Disha Kesharwani

1

, Rishi Paliwal

2

, Trilochan Satapathy

1

, Swarnali Das Paul

3

*

Abstract

Rheumatoid arthritis is a severe autoimmune disor- der, related to joints. It is associated with serious carti- lage destruction. This causes disability and reduces the excellence of life. Numerous treatments are existed to combat this disease, however, they are not very efficient and possess severe side effects, higher doses, and fre- quent administration.

Therefore, newer therapies are developed to overcome all these limitations. These include different monoclonal antibodies, immunoglobulins, small molecules used for immunotherapy and transgenes for gene therapy. One of the main goals of these new generation therapeutics is to address the underlying distressing biological pro- cesses by specifically targeting the causative agents with fewer systemic side effects and greater patient console.

It is very fortuitous that loads of progressive investiga- tions are going on in this field and many of them have entered into the successful clinical trial. But till date, a limited molecule has got FDA clearance and entered the market for treating this devastating disease.

This review highlights the overview of conventional therapy and advancements in newer therapeutics in- cluding immunotherapy and gene therapy for rheu- matoid arthritis. Further, different novel techniques for the delivery of these therapeutics of active and passive targeting are also described.

1. Background

The word arthritis came from the Greek word "for joint inflammation". It mainly affects the joints of the body. But sometimes other tissues of the body, such as the kidneys, eyes, skin, etc. are also getting affected [1]. Arthritis belongs to the category of T cell-mediated autoimmune disorder in which the immune system of the body attacks its own tissues. It is a disease in which the body fails to recognize the self-molecules from for- eign molecules [2].

In rheumatoid arthritis, the immune responses in- fluence the secretion of rheumatoid factors and evoke destruction of cartilage and bones in progression.

Both the environmental factors and genetic factors are implicated in the progress of clinical indication of RA [2-4]. Joint damage occurs due to the auto-re- action of different immune modulators like effector cells and cytokines. It starts at membranes of synovi- um and then progressively attacks the adjacent struc- tures. The activation of dendritic cells, T cells, plasma cells, B cells, mast cells, macrophages, and angiogen- esis cause sinusitis [5-6]. Amongst these, persistently

Review article

Key Words

Arthritis, Cytokines, Disease-modifying anti-rheumat- ic drugs, TNF- α Interleukins, abatacept, rituximab, glucocorticoid.

This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

This paper meets the requirements of KS X ISO 9706, ISO 9706-1994 and ANSI/NISO Z39.48-1992 (Permanence of Paper).

*Corresponding Author

Swarnali Das Paul. Associate Professor, Faculty of Pharmaceutical Sciences, SSTC,SS- GI,Bhilai, Pin: 490 020, C.G, India.

Tel: Tel: +91-997-725-8200 Email: swarnali34@gmail.com

2019 Korean Pharmacopuncture Institute http://www.journal-pharm.com Received: May 12, 2019 Reviewed: Jun 11, 2019 Accepted: Nov 25, 2019

1 Columbia Institute of Pharmacy, Raipur, Chhattisgarh, India

2 Assistant Professor, Faculty of Pharmacy, IGNTU, Amarkantak, Madhya Pradesh, India

3 Associate Professor, Faculty of Pharmaceutical Sciences, SSTC,SSGI,Bhilai,C.G, India

(2)

activated synovial macrophages are one of the leading factors for producing inflammation in RA. Fig. 1 explains the progression of rheumatoid arthritis. However, the in- tensity of inflammation on the joints and degradation of tissues depends on the number and level of macrophage activation[6- 7]. Therefore, over the last few decades, the RA treatment has been progressed by considering its inter- nal mechanisms so that drugs can be developed to target at the molecular level. Table 1 depicts different molecular targets explored for drug targeting [8-11].

These novel targeted drugs have proved the enormous potential for countering disease.

However, distinguished side effects, long-term treat- ment challenges and cost of treatment are till have to be considered. The most important reason behind this is the nonspecific delivery of drug molecules. So nowadays the research is turned towards the development of targeted delivery strategies to the inflamed joints [12]. Although in- tra-articular injection is one of the best options as target- ed therapy, repeated joint needling limits its utility which enhances the risk of infection. As well as it offers the lo- cal administration of the inflamed joints this cannot be a choice in case of polyarthritis and systemic disease [13].

The detailed study of inflammatory diseased tissue shows an abnormal increment of macrophages and other cellular factors. This leads to enhancing the targeting of therapeu- tics by different novel delivery techniques. These targeted delivery systems accumulate passively into inflamed tis- sues via different mechanisms like the enhanced permea- bility and retention, surface conjugation with a ligand and others. Surface conjugation helps in the active attachment

to receptors which are proliferated by affected cells so that systemic side-effects can be reduced and efficacy can be increased[14-15].

In this review, the overview of conventional drugs for RA and advancements in the newer therapies as medical op- tions for management are discussed. Clinical trial updates of different treatment are also reviewed and special con- sideration is given to different novel delivery systems for their delivery.

2. OVERVIEW OF CONVENTIONAL RA THER- APEUTICS

The present-day treatment for RA can be classified into four classes:

i. Non-steroidal anti-inflammatory drugs (NSAIDs) ii. Glucocorticoids

iii. Non-biologic Disease Modifying Anti-rheumatic Drugs (DMARDs)

iv. Biologic Disease Modifying Anti-rheumatic Drugs (DMARDs)

These recognized therapies are summarized in Table 2.

Under each section the mechanism of action, side effects, dose and new drug candidates of each class of the drug.

Different NSAIDs are generally the first choice of drugs for the treatment of RA as well as for osteoarthritis and other musculoskeletal disorders. They are generally used for the symptomatic relief of the disease and do not have any ef- fect on the cause of the disease. These drugs reduce pain and swelling by inhibiting the cyclooxygenase enzyme which is responsible for inflammation. Cyclooxygenase -1 and Cyclooxygenase -2 are the two isoforms of the enzyme [16-17]. Corticosteroids are the second leading prescribed drug for the treatment of RA. These agents possess strong anti-inflammatory effect as well as analgesic effect. An es- tablished Guideline for RA known as “National Institute for Health and Clinical Excellence (NICE)” states that cor- ticosteroids should be used only after the application of all other treatment options and after gaining the complete knowledge about the side effects as these drugs produce severe osteoporosis and fractures, Cushing's syndrome, weight gain, cataract, hypertension, etc. [18-19].

DMARDs are the drug of choice for RA treatment. These drugs mainly work on the progression of the disease, hence agents prevent further joint destruction and loss. This ther- apy has been found successful in many cases. It eliminates the need for any other treatment. Other drugs can be used for symptomatic relief until the completion of therapy [20- 22]. Mainly the biologic and non-biologic disease modi- fying anti-rheumatic drugs (DMARDs) are the important pharmacological therapies for RA. DMARDs of non-bi- ologic origin is also popular as small molecule DMARDs or low molecular weight DMARDs and a wide variety of chemically diverse drugs are in this group. These drugs are further divided into two classes: traditional and novel.

In this section, only traditional DMARDs will be discussed.

Traditionally DMARDs have been used since the 1920s are Figure 1

(3)

5 S. No. Molecular

Targets Role Occurrence Example

of Targeting Drugs

1. Cyclooxygenase pathway

Biosynthesis of prostanoid, biologically active substances, involved in pathological conditions inflammation.

Cytosol and tissue

Celecoxib, Piroxicam, Naproxen, Valdecoxib

2. Tumor Necrosis Factor-α

Activation of macrophages, synovial fibroblasts, endothelial cells, MMPs and adhesion molecule expression and release of other cytokines and PGs.

Synovial fluid and tissue

Infliximab, Etanercept, Adalimumab, Golimumab, Certolizumab pegol

3. Interleukin-1

Potent inducer of MMPs, eicosanoids, and receptor activator of NF- κB Ligand, Hyaline cartilage synthesis inhibitor.

Synovium Anakinra

4. Interleukin- 6 Activation of oesteoclasts, bone resorption, upregulates intercellular cell adhesion molecules 1 expression.

Serum and

synovial fluid Tocilizumab, lactoferin

5. Interleukin- 8 -- Synovium ABX-IL8

6. Interleukin- 10 Inhibit the production of cytokines and Enhancement of production of IL-1RA

Synovial tissue 7. Interleukin-12 Act in synergy with anti-TNF-α antibodies Synovial fluid

ABT-874

8. Interleukin-15 Activates T-cells, Stimulation of macrophages to release TNF-alpha

Joint

Synovium HuMax-IL-15 9. Interleukin-

17Alpha

Activation of IL-1, 6 and 8, implicated in osteoclast

activation causing bone resorption in RA Synovium -- 10. Interleukin-18 IL-1 and TNF production enhancement Synovium IL-18bp

11. Matrix

Metalloproteinase Involved in bone and cartilage degradation Joint

Synovium Trocade (Ro 32-3555) 12. Nuclear

Factor-κB Cytosol Iguratimod

13. Cathepsin- B Cleaves aggrecan and enhancement of RA Synovial

tissue --

14. Aggrecan Maintainance of cartilage integrity Synovium --

6 15. Osteopontin Stimulates cell adhesion, migration, and specific

signaling function.

Extracellular fluid, and inflammation site

--

16. Prostaglandin

(PG) Bone resorption stimulator Osteocyte Celecoxib, Piroxicam,

Naproxen, Valdecoxib 17. P38MAPKs Inhibition affects TNF production Synovial

tissue

Pamapimod, VX-702 and SCIO-469 18. Oncostatin M Synergistic with IL-1, promote cartilage damage Synovial

fibroblasts -- 19. Collagen I Osteoblastic differentiation of the bone marrow cells Bone cell -- 20. Collagen II Maintain the integrity of cartilage Cartilage -- 21. T lymphocyte essential for the continued activity of inflammation in

RA Thymus Abatacept

22. B lymphocyte Antigen presentation

Secretin of pro-inflammatory cytokines

Bone marrow, synovial membrane

Rituximab

23. Janus Kinase (JAK)

affect intracellular signaling through their association with transcription factors known as STATs Synovium

Tofacitinib, VX-509, Baricitinib (formerly LY3009104/

INCB028050), Ruxolitinib (formerly INCB018424) 24. Spleen Tyrosine

Kinase (Syk)

Syk is theoretically connected to inflammation and bone resorption.

Fostamatinib (formerly R406; R788 is the prodrug),

2. OVERVIEW OF CONVENTIONAL RA THERAPEUTICS The present-day treatment for RA can be classified into four classes:

i. Non-steroidal anti-inflammatory drugs (NSAIDs) ii. Glucocorticoids

iii. Non-biologic Disease Modifying Anti-rheumatic Drugs (DMARDs) iv. Biologic Disease Modifying Anti-rheumatic Drugs (DMARDs)

Table 1 Molecular Targets in Rheumatoid Arthritis

(4)

8 S. No. Category Drug Brand name Adult

Dose Mechanism Side effects Company FDA Approval

1. NSAIDs

Celecoxib Celebrex

Tablet 100 to 200 mg

orally twice daily.

COX-1 and COX-2 inhibitor

Dyspepsia, peptic ulcer disease, bleeding, myocardial

infarction, Increased Blood pressure, Heart failure.

G.D. Searle FDA 1998

Piroxicam Feldene 20-4-mg

daily Pfizer FDA 1982

Nabumeton

e Relafen 1000 mg

daily

Teva Pharmaceutic

als Ltd. FDA 2000 Naproxen

(i) Anaprox (ii) Neprelan

500- 1000 mg daily

375mg, 500 mg daily

Atnahs Pharma Inc.

FDA 1980

Etodolac Lodine

100- 200 mg not more than 1000mg/d

ay

Teva FDA 2000

Rofecoxib Vioxx 25 mg

Daily Merck FDA 2002

Valdecoxib Bextra Pharmacia,

Pfizer FDA 2001 Ibuprofen

Famotidine and Duexis

mg/26.6 800

mg Horizone

Pharma FDA 2011

2. Glucocorti coids

Prednisone Deltasone, Liquid Pred,

Sterapred

5- 60 mg/day in

divided doses

Inhibition of macrophage accumulation,

reduction of capillary permeability

Osteoporosis Stomach

ulcer Increased

blood pressure Irritability

and/or excitability

Increased blood sugar/glucose

Cataracts (clouding of

eye lenses)

Pharmacia

and UP John FDA 1955 Methyl

Prednisone

Depopred, Medrol, Methacort, Predacorten

2-60 mg/day

Hydrocorti

sone A-Hydrocort,

Cortef 10-320

mg/day Hospira FDA 2006

Dexametha sone

Decadron, Dexpak, Hexadrol, Taperpak

0.75 – 9

mg/day Merck FDA 1958

3. Nonbiologic DMARDs

Methotrexa

te (i) Trexall (ii) Xatmep

Maximum weekly dose: 20

mg.

Inhibition of aminoimidazol e carboxamide ribonucleotide

(AICAR) transformylase

thymidylate and synthetase

Nausea, mouth ulcer,

hair loss, cytopaenias, elevated liver

enzymes, rarely pneumonitis

Silvergate Pharms

(i) FDA 2001 (ii) FDA

2017 Table 2 Overview of Conventional RA Therapeutics

(5)

mainly non-biologic. They are including Sulphasalazine, Chloroquine, Methotrexate, Azathioprine, Cyclosporine A, Minocycline, and others. Even in the new era of biolog- ic, methotrexate (MTX) still remains the drug of choice for RA and prepared as a standard to probate many other new disease-modifying drugs.

Novel Non-biologic DMARDs are further divided into four classes: phosphoinositide 3-kinases (PI3Ks) inhib- itors, glycosidase inhibitors, matrix metalloproteinases (MMPs) inhibitors and cathepsin inhibitors [23].

2.1. Current FDA status of conventional RA therapeutics

It is very unfortuitous that till 2017 only 25 (approx) drugs got approval for the RA treatment by the Food and Drug Administration (FDA). These drugs include corticoster- oids, NSAIDs, different antibodies, and immunoglobulins.

Methotrexate is one of the prime and vintage RA drugs of the mainstays for its dual capacity to reduce pain-swelling and modify joint damage to prevent disease progression over time. Methotrexate was first applied in 1947 as folate antagonist in pediatric leukemia. In 1951, its effect on RA was established by Gubner and it got approved by the FDA in 1988. In the last 25 years, this drug has become a prima- ry standard in the treatment of adult RA. Another pioneer drug prednisolone or methylprednisolone was the drug of

8 S. No. Category Drug Brand name Adult

Dose Mechanism Side effects Company FDA Approval

1. NSAIDs

Celecoxib Celebrex

Tablet 100 to 200 mg

orally twice daily.

COX-1 and COX-2 inhibitor

Dyspepsia, peptic ulcer disease, bleeding, myocardial

infarction, Increased Blood pressure, Heart failure.

G.D. Searle FDA 1998

Piroxicam Feldene 20-4-mg

daily Pfizer FDA 1982

Nabumeton

e Relafen 1000 mg

daily

Teva Pharmaceutic

als Ltd. FDA 2000 Naproxen

(i) Anaprox (ii) Neprelan

500- 1000 mg daily

375mg, 500 mg daily

Atnahs Pharma Inc.

FDA 1980

Etodolac Lodine

100- 200 mg not more than 1000mg/d

ay

Teva FDA 2000

Rofecoxib Vioxx 25 mg

Daily Merck FDA 2002

Valdecoxib Bextra Pharmacia,

Pfizer FDA 2001 Ibuprofen

Famotidine and Duexis

mg/26.6 800

mg Horizone

Pharma FDA 2011

2. Glucocorti coids

Prednisone Deltasone, Liquid Pred,

Sterapred

5- 60 mg/day in

divided doses

Inhibition of macrophage accumulation,

reduction of capillary permeability

Osteoporosis Stomach

ulcer Increased

blood pressure Irritability

and/or excitability

Increased blood sugar/glucose

Cataracts (clouding of

eye lenses)

Pharmacia

and UP John FDA 1955 Methyl

Prednisone

Depopred, Medrol, Methacort, Predacorten

2-60 mg/day

Hydrocorti

sone A-Hydrocort,

Cortef 10-320

mg/day Hospira FDA 2006

Dexametha sone

Decadron, Dexpak, Hexadrol,

Taperpak

0.75 – 9

mg/day Merck FDA 1958

3. Nonbiologic DMARDs

Methotrexa

te (i) Trexall (ii) Xatmep

Maximum weekly dose: 20

mg.

Inhibition of aminoimidazol e carboxamide ribonucleotide

(AICAR) transformylase

thymidylate and synthetase

Nausea, mouth ulcer,

hair loss, cytopaenias, elevated liver

enzymes, rarely pneumonitis

Silvergate Pharms

(i) FDA 2001 (ii) FDA

2017

9 2.1. Current FDA status of conventional RA therapeutics

It is very unfortuitous that till 2017 only 25 (approx) drugs got approval for the RA treatment by the Food and Drug Administration (FDA). These drugs include corticosteroids, NSAIDs, different antibodies, and immunoglobulins. Methotrexate is one of the prime and vintage RA drugs of the mainstays for its dual capacity to reduce pain-swelling and modify joint damage to prevent disease progression over time.

Methotrexate was first applied in 1947 as folate antagonist in pediatric leukemia. In 1951, its effect on RA was established by Gubner and it got approved by the FDA in 1988. In the last 25 years, this drug has become a primary standard in the treatment of adult RA. Another pioneer drug prednisolone or methylprednisolone was the drug of choice in RA in most chronic cases and to date, they are in the system.

The FDA approved methylprednisolone in October 1957 for treatment of RA [24].

Leflunomid

e Arava

Initial dose: 100 mg orally once a day for 3 days

Inhibits T-cell proliferation production of and autoantibodies by B cells.

disturbance, GI hair loss, weight loss, rash and itch,

mouth ulcer, headache, cytopaenias, hypertension

Sanofi

Aventis US FDA 1998

Sulfasalazi

ne Azulfidine 0.5-

2gm/day

Inhibits the release of inflammatory

cytokines,

Nausea, abdominal

pain, hair loss, cytopaenias, elevated liver

enzymes, agranulocytos

is, skin rashes.

Pharmacia &

Upjohn FDA 1996

4. Biologic DMARDs

Tocilizuma

b Actemra

Dose - 4 mg/kg followed

by an increase to

8 mg/kg Inhibits Interleukin – 6

receptor

Upper respiratory

tract infections, nasopharyngit

is, headache, hypertension, increased AL, injection site

reactions

Genentech FDA 2017

Golimuma

b Simponi Dose-

50mg Centocor

Ortho

Biotech FDA 2009

choice in RA in most chronic cases and to date, they are in the system. The FDA approved methylprednisolone in October 1957 for treatment of RA [24].

The approval of Lodine by Wyeth containing etodolac was a remarkable confrontation in the year 1996. Immediately after that Azulfidine enteric coated tablets of sulfasalazine and Naprelen containing naproxen came into the market in the same year. However, sulfasalazine was permitted for medical application in 1950 in the United States. Initially, this drug was formulated as an enteric-coated formulation marketed as AZULFIDINE EN-tabs. These tablets were specifically intended to control nausea and stomach up- set. Till date, this tablet is the only FDA approved formula- tion of sulfasalazine for the treatment of both juvenile and adult RA [25-27].

In the year of 1997, the first generic equivalent of Lodine came into the market just after one year by Royce Labo- ratories was Etodolac. Another three NSAIDS got approv- al in this year: Arthrotec by Searle, Ketoprofen by Schein Pharmaceutical which is a generic equivalent or Oruvail and Tolmetin Sodium by Teva Pharmaceutical [28-29].

Hoechst Marion Roussel company launched Arava con- taining Leflunomide tablet in 1998. Leflunomide is an immunosuppressive disease-modifying antiarthritic drug, used in RA and psoriatic arthritis [30]. In 2001 Valdecox- ib (NSAIDS) tablets under the brand name Bextra got ap- proval launched by Pharmacia Pfizer. However, because of the increased risk of heart attack and stroke, it was re- moved from the U.S. market in 2005. The same incidence

(6)

also happened with Vioxx containing rofecoxib drug (ap- proved April 2002, withdrawn October 2004) [31-32].

After a long gap in 2010, Vimovo a combination product of naproxen and esomeprazole by AstraZeneca got per- mission for RA in patients at threat for NSAID linked ulcers [33]. In 2011, another combination product of ibuprofen and famotidine got approved under the name Duexis for the relief of RA as well as osteoarthritis by Horizon Phar- ma. It was also indicated for gastric ulcers associated with therapy. By this time, many monoclonal antibodies, im- munoglobulins got the green signal from the FDA for treat- ing this devastating disease. Immediately after one year, in 2012 delayed-release tablets of prednisone (Rayos) get underway by Horizon Pharma for curing different inflam- matory diseases, including rheumatoid arthritis, psoriatic conditions, COPD, and asthma [34].

3. ADVANCES IN NEW GENERATION RA THERAPEUTICS

The autoimmune characteristics of RA which is primarily evident as ‘chronic inflammatory arthropathy', create this treatment challenging for years. Conventional drugs have no (NSAIDs) or limited (Corticosteroid) authority on this autoimmune nature of the disease. Moreover, corticoids and other non-biologic DMARDs are inadequate and only capable of achieving clinical remission in combination.

Further, they possess common unwanted side effects such as stomach upset, nausea, vomiting, diarrhea, liver prob- lems, etc. In addition to this, approximately half of the RA patients don't show any promising reaction to traditional DMARDs [21].

Thus, one of the foremost goals of new generation RA ther- apeutics is to repress the fundamental biological processes which cause bone erosion, joint destruction, and progres- sion of physical comorbidities. These medicines have a fast therapeutic action and possess comparatively less system- ic side effects than former treatments. Most significantly, these therapeutic substances can modify the fundamental cause of inflammation and cell damage. These amazing therapeutic agents are including biologic DMARDs or bi- otech drugs used in immune therapy and specific trans- genes for gene therapy [21]. In this section, introduction, advancements, clinical aspects and other details of these newly introduced agents have been discussed.

3.1. Immunotherapy for RA

The amendment of immune therapies as especially do target the molecules and cells accountable within the immunopathogenesis is very essential for RA treatment.

The preceding endeavors at RA immunotherapy had been cells targeted rather than molecules. The detected T lym- phocyte values into the investiture about RA resulted in endeavors according to alternate the immune replication by denotes of depleting T lymphocytes [35]. However, this method was once generally unfruitful. It was found rational to aim at T cells, so those hold a sanctioned role

among initiates or directing immune replications. During this length, many other researchers have been focused on provocative cytokines, along a most important attempt fo- cused regarding the provocative TNF-α. At the beginning of the 1980s, TNF-α was recognized in the synovial film of RA patients. 27 years later in 1980s new TNF-α blockers had been produced. The drug named CA2 (infliximab) was first in this category.

Last study of few years shows that many patients impas- sive to established therapies have proved effective in treat- ment with novel biological agents. The rationale behind this may be the efficiency of highly specific targeting ap- proach to provocative cytokines and other involved cells and along with their surface molecules in the RA patho- genesis. They mainly act by three mechanisms:

First, prevention of binding of the cytokines to its cell-sur- face receptors in different ways, including monoclonal antibodies, soluble receptors, and natural antagonists.

Second, inhibition of production/proliferation of provoc- ative cytokines by anti-inflammatory cytokines such as in- terleukin-4, 10 or 13. Third, exclusion of the inflammatory cells or interference with cell function by targeting either biological agent against differentiation or cell-surface an- tigens attached functionally.

Presently used biologic DMARDs are including TNF an- tagonists (etanercept, infliximab, golimumab, adalimum- ab), IL-1and IL-6 inhibitor (tocilizumab), T cell inhibi- tors(abatacept), B cell inhibitors, anti CD20 (Rituximab).

However, latest biologic DMARDs under investigation are phospholipase A2 (PLA2) inhibitors, vascular endothelial growth factor (VEGF) inhibitors, chemokines blocker, col- ony stimulating factor” (CSF) inhibitors and others [36].

Amongst recently used DMARDs TNF blockers are much more extravagant and consequently payers endeavor to re- strain their exploitation of patients whose disease cannot otherwise be controlled. Therefore, traditional therapy is a junction point in RA treatment where cost considerations become very authentic. While biologic therapies are effi- cacious, they are sumptuous. The alternative approach of amalgamation therapy with conventional DMARDs, such as hydroxychloroquine and sulfasalazine together with methotrexate (soi-disant "triple therapy") and the utiliza- tion of glucocorticoids as bridge therapy, is additionally ef- ficacious. It is much less extravagant, but it involves several medicines and the insertion of glucocorticoids and their associated side effects [37].

3.1.1. Current FDA status of immune-ther- apeutics

Table 3 depicts the FDA approved immune-therapies for RA with their mechanism and side effects. Initially, Enbrel (Etanercept) was launched in November 1998. It was the first targeted biologic for RA. It reduces the symptoms and inhibits the progression of structural damage in patients facing moderate to highly active rheumatoid arthritis. It is also useful in juvenile polyarticular type RA. It is basical- ly a TNF antagonist. Further, in January 2002, it received approval for diminishing the symptoms of arthritis in pa-

(7)

15 S. No. Approval year Brand

name Biologics Company Dosage

form Mechanism Side effects 1. November 1997 Rituxan Rituximab Genentech IV solution antiCD20

antibody Skin Rashes

2. November

1998 Enbrel Etanercept Immunex

Corporation SC injection TNF inhibitors

Serious infections including tuberculosis, upper

UTI

3. November

2001 Kineret Anakinra Amgen Injectable IL-1 receptor antagonist

Diarrhea, stomach pain, Headache 4. June 2002 Remica

de Infliximab Centocor Ortho Biotech

Intravenous

infusion TNF inhibitors Chest pain, sore throat, dizziness,

fatigue

5. December 2002 Humira Adalimumab Abbott

Laboratories SC injection TNF inhibitors

Bruising at the injection site, upper

respiratory infection, nausea

6. December 2005 Orencia Abatacept Bristol- Myers

Squibb SC injection CTLA4-Ig fusion protein

Increase risk of serious infections,

Skin irritation, itching rashes, swelling and pain

7. April 2009 Simponi Golimumab Centocor Ortho

Biotech SC injection TNF inhibitors

Increase the risk of serious infections

including tuberculosis,

8. May of 2009 Cimzia Certolizumab pegol UCB SC injection TNF inhibitors Tuberculosis, UTI, headache

9. June 2009 Ilaris Canakinuma

b Novartis Powder for

injection

anti-IL-1β receptor antibody

Bronchitis, Diarrhoea, Gastroenteritis, vertigo, weight

increase 10. September

2009 Stelara Ustekinuma

b Janssen

Biotech SC injection Human IgG1k monoclonal

antibody

Nasopharyngitis, diarrhea, upper UTI,

nausea

11. January

2010 Actemr

a Tocilizumab Genentech IV infusion anti-IL-6 receptor antibody

Serious infections, elevated liver

enzymes neutropenia, decreased platelet

counts

12. November

2012 Xeljanz Tofacitinib Pfizer Tablet, extended

release

Janus Kinase inhibitor

Upper respiratory tract infection,

Diarrhoea, Nasopharyngitis 13. March 2014 Otezla apremilast Celgene Tablet PDE4

inhibitor Diarrhoea, nausea, headache 14. March

2016 Taltz Ixekizumab Eli Lilly SC injection solution

Anti IL- 17A receptor antibody

Injection site reactions, tinea infection, nausea

15. May 2017 Kevzara sarilumab Sanofi SC injection IL- 6 receptor antagonist

Neutropenia, increased ALT,

injection site erythema, UTI Table 3 FDA Approved Immunotherapeutic For Treatment Of RA

(8)

tients with psoriatic arthritis for which it’s the first therapy [38].

Studies reported that TNF blockade is one of the most ef- fective methods for RA treatment. It leads to the evolution of distinct inhibitors of TNF like Infliximab, Adalimumab, Certolizumab pegol, Golimumab. In June 2002, infliximab (IFX) came into the market. It is a chimeric monoclonal antibody (mAb) against TNF-α [39]. After six months an- other TNF-α blocker, Humira (adalimumab) by Abbott Laboratories got approval.

Anakinra (brand name Kineret) the first biopharmaceu- tical drug used to treat rheumatoid arthritis got approval in November 2001. It is a recombinant human receptor antagonist acting on IL- 1 and nonglycosylated form of protein.

A specific co-stimulation modulator named Abatacept is accepted in 2005. It binds to CD80/86 and inhibits T-cell activation and thus it modulates its interaction with CD28 which initiates, the co-stimulatory signal required for the T cell activation. This drug is suggested for patients not re- sponding to different DMARDs or TNF inhibitors, takes an approach that differs from the TNF inhibitors [40].

It is notable that amongst these immune-therapeutics, etanercept, adalimumab, and anakinra are self-injecta- ble drugs, while infliximab, rituximab, and abatacept are infused products [41]. A product of Novartis, Canakinum- ab (ACZ885, Ilaris) acts by neutralizing 1β signaling. It is a human anti-IL-1β monoclonal antibody. It suppresses inflammation in patients with autoimmune disorders [42]. STELARA® is a human IgG1k monoclonal antibody.

It attaches specifically to the protein on p40 subunit and utilized by both the cytokines interleukin -12 and 23. STE- LARA® (ustekinumab) is prescribed to the patients of ac- tive psoriatic arthritis. A combined form of it and metho- trexate is also frequently suggested [43].

Tocilizumab denoted as TCZ was the first biological DMARD and it targets to IL-6. The clinical trials for oth- er inhibitors of Interleukin-6 are also in progress. These include sarilumab the monoclonal antibody (REGN88/

SAR153191), and nanobody (ALX-0061), sirukumab, olokizumab, and MEDI5117. EVZARA targets and binds with high affinity to IL-6 receptors (sIL-6R and mIL-6R) came into the market in 2017 [44].

Janus kinase pathway inhibition is an advanced mecha- nism. It inhibits the intracellular effects of several inflam- matory cytokines and completely modulates the immune and inflammatory response. Tofacitinib is the first oral non-biologic DMARD, approved in 2012. Baricitinib is an- other one from the same group. For the patients with an unsuccessful DMARD history tofacitinib is the best drug [45].

3.2. Gene therapy for RA

In the understanding of the molecular mechanisms be- hind RA pathogenesis cause a continuous advancement over the past years. It results in the development of many kinds of targets of novel gene therapy. It is an established fact that RA pathogenesis is involved with activation of specific T cells, osteoclasts, monocytes, fibroblasts, B

cells, endothelial cells and macrophages [46]. Along with this excess production of pro-inflammatory factors like cytokines and chemokines, rheumatoid factor and ma- trix metalloproteinases are also responsible. Each of these factors can be manipulated as a target in multiple ways using gene therapy. This section presents a review of the latest advances in gene therapy for RA treatment.

Therapeutic genes are generally delivered by two meth- ods: in vivo and ex vivo. These methods can accomplish the need of local as well as systemic delivery of genetic material. Generally, the intraarticular route is used for gene transfer in local in vivo strategies [47]. However, ex vivo strategies include three steps:

i. Exclusion of synoviocytes from the affected area usually joint

ii. In- vitro Transduction

iii. Transduced cells are re-injected into the joints.

The systemic gene therapy consists of the transfer of genes to the cells and these cells synthesize the gene product. These gene products are then secreted into the circulation. This approach is beneficial as rheumatoid arthritis has its systemic nature and it has the capacity to target more than one joint simultaneously. This also leads to non-specific immune suppression with increased sus- ceptibility to infection [48]. Many transgenes are available which are utilized for RA in different methods some of the important examples are summarized here.

In a report ‘IL-1 Ra' transgene was introduced by naked DNA method to DBA/1 mice. Arthritis was induced by using an adjuvant-induced arthritis method. Four sites of hind limbs were injected intramuscularly. The results depicted that the onset of collagen-induced arthritis was completely prevented with decreased synovial inflamma- tion, cartilage destruction, with a lesser expression of IL- 1β in ankle joints [49].

One more study administered ‘IL-1 Ra co-expressed with GFP’ by i.e. injection into mice via adenovirus. The results reveal the severity of arthritis was reduced along with re- duced foot-pad swelling, inflammatory cell infiltration, and synovial proliferation. Additionally, it increased Th1 driven IgG2a antibodies and Th2driven IgG1 antibody and conserved the proteoglycan concentration [50]. It was observed that impediment in enduring provocative autoimmune disease after intravascular tail injection of a single-chain antibody into ‘DBA/1 LacJ mice [51]. A study on ‘adoptive cellular gene therapy’ where T-cell was hy- brid and transduced with RT administered CIA by intra- venous tail injection. They reported transgene expression was constrained to the paw and decreased the level of IL-6 [52]. One more study conducted on both IL-1Ra and SiL-1RAcP. They were injected CIA by i.v. Via adenovirus into DBA/1 mice. The result disclosed sIL-1RAcP refur- bished CIA without affecting T cell immunity, whereas IL-1Ra refurbished CIA and repressed lymphocyte prop- agation [53]. In a motivating study, researchers used var- iants of hTNFR-Is as monomeric, dimeric and ‘chimeric hTNFRIs/mIgG1’ type. These were introduced via ET of plasmids DBA/1 mice, CIA by intramuscular injection.

Researchers found hTNFR-Is/mIgG1 at the beginning of CIA condensed clinical and histological manifestations of the disease. Dimeric form demonstrated the effective-

(9)

ness of local expression in a dose-dependent manner and lasted a minimum of 6 months. Researchers found after intraarticular injection of an adenovirus (over-expressing IL-18BP) to mouse knees before the sign of CIA offer many favorable effects on the course of RA disease. These in- clude the control incidence of bone damage, occurrence of disease, and collagen II-specific IgG2a antibody levels [54].

In RA patients 08 clinical trials of gene therapy have been initiated. One of these clinical trials was closed that was aimed to conduct genetic synovectomy, due to inade- quate enrollment. The only clinical trial data on rheuma- toid arthritis have published by Evans and coworkers. It came from a phase I protocol of arthritic patients. This data approved the safety and ease of gene transfer to ar- thritic joints [55].

4. ADVANCES IN DRUG DELIVERY SYSTEMS FOR THE RA THERAPEUTICS

Despite the presence of a wide variety of therapeutics for RA, the major challenge lies in their successful delivery to the affected area. Along with this, most of these therapeu- tics often cause side effects and drug resistance. The most important reason behind this is the nonspecific delivery of drug molecules. So nowadays the research is turned towards the development of targeted delivery strategies to the inflamed joints [41]. The changed properties of the inflammatory site, such as a change in pH, temperature, EPR effect and overexpression of various cells have the greatest potential for targeting. Therefore, different deliv- ery systems are establishing their arena by targeting the drug to the site, reducing the amount of drug and adverse effect [56]. Fig. 2 shows some novel carriers used and fig.

3 illustrates the mechanism of different newer delivery systems for targeting RA. Advancements of these delivery systems and their efficacy in carrying the therapeutic mol- ecule to the target site are described here.

Figure 2

Figure 3

4.1. Nanoparticles

Nanoparticles are the particles microscopic in size and smaller than 1micron. They generally measure approxi- mately 1 - 1000 nm in size and differ their properties from their macroscale forms. The drug can be embedded in the nanoparticles matrix or it can be adsorbed onto the surface. Nanoparticles also enhance the solubility and bioavailability of poorly soluble drugs. These nanocarrier systems also have properties of the high surface to volume ratio, enhanced permeability and retention effect, sus- tained action, etc. similar to other nanocarriers [56-57].

Due to these unique properties, they are successfully used for drug targeting in different diseases including RA.

Solid Lipid Nanoparticles (SLN) are suitable colloidal carriers. These are used for delivery of poorly soluble drugs. These colloidal carriers are differentiated from na- nostructured lipid carriers (NLC) by the composition of the solid matrix. They are alternative carriers to liposomes and emulsions. As the SLN are prepared from biocompat- ible and biodegradable lipids, these systems are highly acceptable and tolerable. The lipid nanoparticles are the safest nano-carrier system [58].

Lee et al., 2013 have developed half-shelled gold nano- particles attached with Arginine- Glycine- Aspartic pep- tide (RGD) for RA treatment. The active constituent was methotrexate. Methotrexate belongs to DMARDs class of antirheumatic drugs. RGD peptide was used as a ligand for targeting the inflamed tissue. They concluded that the prepared nanocarrier system minimized the adverse ef- fects of methotrexate and enhanced its efficacy. On the ba- sis of obtaining results, they suggested that this nano-car- rier can also be used for the delivery of other DMARDs efficiently [59]. Satya Prasad et al., 2016 explained that multifunctional nanoparticles are used for Rheumatoid arthritis or other inflammatory diseases. They described the Zinc oxide and Zinc sulfate nanoparticles, gold nano compounds such as auranofin, aurothioglucose, silver nanoparticles, and metallic copper nanoparticles were used to improve the clinical status of arthritic patients.

(10)

They concluded that GOLD nanoparticles are the best remedy for rheumatoid arthritis [60]. Hwiwon Lee et al., 2014 prepared hyaluronate gold nanoparticles and Tocili- zumab complex for rheumatoid arthritis treatment. Gold nanoparticles are used as a carrier with an antiangiogenic effect. The effect of the complex was approved by ELISA and western blot analysis in mice using adjuvant-induced arthritis [61]. Homma et al., 2010 told that Hyaluronic Acid (HA)-conjugated methotrexate (MTX) nanoparticles reduced the proliferation of fibroblast-like synoviocytes (FLs) in vitro, and symptoms of arthritis were consequent- ly alleviated [62]. Zheng et al., 2015 successfully prepared oil, water nanoemulsion of curcumin for improving the low oral bioavailability of curcumin, which can facilitate the formulation of oral dosage forms [63]. Shailaja A.K.

et al., 2016 prepared polymeric nanoparticles containing Mefenamic acid. They used the ionotropic gelation tech- nique. After the development of the nanoparticles, they characterized these nanocarriers and found that these systems possessed excellent properties such as less par- ticle size, greater stability and controlled drug release for 12 hours [64]. Pang-hu Zhoua et al., 2018, synthesized hyaluronic acid-chitosan nanoparticles containing plas- mid DNA encoding Cytokine response modifier A (HA/

CS-CrmA). They used the method of complex coacerva- tion of cationic polymers. The nanoparticles qualified dif- ferent evaluation parameters. The result showed that the prepared nanoparticles safely transfected arthritic cells and released the drug with sustained effect. It can be con- cluded that the prepared nanoparticles prevent cartilage destruction and inflammation [65]. Wenshuai Fan et al., 2018, used kartogenin (KGN) molecule that is reported to show a protective and regenerative effect on cartilage, and polyurethane nanoparticles were prepared. The kar- togenin conjugated polyurethane nanoparticles showed a sustained release of drug with regular spherical shape and size. The intra-articular (IA) injection of KGN conju- gated polyurethane nanoparticles proved efficacious with less cartilage degeneration as compared to IA injection of plane KGN [66]. Jiesheng Ye <http://www.sciencedi- rect.com/science/article/pii/S0378517307008435> et al., 2008, studied in detail about SLNs loaded with actarit and incorporated in the intravenous injection formulation.

Actarit is an anti-rheumatic drug with a poor water-solu- bility profile.

To improve the therapeutic efficacy of the poorly sol- uble drug and to reduce its nephrotoxicity and GIT dis- turbances were the primary objectives of the study. They concluded that actarit SLN in the form of injectable was promising passive targeting therapeutic agent for RA [67].

Antônio Luiz Boechat et al., 2015 developed MTX-Lipid core nanocapsules and MTX solution. They evaluated their efficiency for the reduction of inflammation. MTX- LNC were shown better results than the MTX solution for reducing different inflammatory agents. Along with this, they found that the nanocarrier system has reduced the dose of MTX. From the found results, they concluded that the LNC containing MTX are a very propitious system for treatment of inflammatory disorders [68]. Meiling Zhoua et al., 2018, developed a targeted system of solid lipid na- noparticle (SLN) containing glucocorticoid prednisolone.

The SLN particles were coated with Hyaluronic acid (HA) shown as HA-SLNs/PD. Hyaluronic receptor CD44 found over-expressed on the surface of synovial lymphocytes, macrophages and fibroblasts in arthritic inflamed joints and HA efficiently binds to these receptors. The results showed that prepared nanoparticles (HA-SLNs/PD) accu- mulated and persisted longer in circulation. The formu- lation was found better in efficacy and safer than the free drug and from SLN of a drug without coating for the treat- ment of inflammatory disorders [69].

4.2. Liposomes

Liposomes are extensively used as drug carriers for rheu- matoid arthritis treatment. They are basically vesicular concentric lipid bilayers. They enclose hydrophilic com- ponents inside them. They are highly biocompatible, bi- odegradable poses less toxicity and commercially impor- tant as they have the ability to entrap both the lipophilic and hydrophilic drugs [70].

Many drugs used for the treatment that RA possesses low bioavailability, limited selectivity, high clearance, etc. And that’s why to require frequent and high dosing to main- tain the therapeutic effect. High doses are also the cause of side effects. Liposomes are the solution for these draw- backs. Weak pharmacokinetic profiles of many drugs can be improved by entrapping the drug in liposomes. They enhance the drug absorption too. Different types of li- posomes possess different specific properties like large liposomes to show enhanced retention, small liposomes are good for passive targeting and PEGylated liposomes also enhance circulation time by reducing the uptake by the liver and spleen. By all these actions the localization of liposomes enhances by enhanced permeability and re- tention (EPR) effect [70-71]. William et al., 1995 developed liposomes containing methotrexate and compared its ef- ficacy with the free methotrexate. They found that meth- otrexate containing liposomes have shown a significant effect on established arthritis. From the obtained results it can be concluded that the liposomal preparations show better results than the free drug [72]. Metselaar et al., 2003 developed PEG encapsulated liposomes containing glu- cocorticoids.

They followed single intravenous treatment and observed its effect on both the joint inflammation and cartilage de- struction. They have also studied the targeting of these for- mulations to the inflammatory site. They concluded that the anti-inflammatory activity of glucocorticoids can be strongly enhanced by the development of long-circulating liposomes [73]. Prabhu et al., 2012 prepared and evaluate the methotrexate nano lipid vesicles for their antirheu- matic activity. They successfully prepared methotrexate nano lipid vesicles with high efficiency, selectivity, and reduced toxicity [74]. Gottschalk et al., 2015 developed cationic liposomes containing MTX and compared their impact with the free MTX. They found that the liposomal MTX showed promising effects on inflamed sites than the free MTX [75]. Rahman et al., 2016 explained that over the conventional carriers, liposomal systems have many ad- vantages related to the delivery of drugs, including merits

(11)

in both passive and active targeting of drug molecules to the inflammatory sites [76]. Mengdi Jia et al., 2018 devel- oped liposomal carriers to deliver dexamethasone and 1,1’-dioctadecyl-3,3,3’,3’-tetramethylindodicarbocyanine by thin film hydration technique. The liposomal carriers for both the molecules were proved efficacious on the basis of results obtained. Both the formulation, taken to- gether, a safe liposomal delivery system was developed to achieve inflammation targeted therapy against arthritis [77].

4.3. Polymeric micelles

These are self-assembled amphiphilic block polymers.

They are biocompatible and biodegradable. They pos- sess a hydrophilic shell and a hydrophobic core. The shell material generally utilized is polyethylene glycol. When polymeric micelles are compared with free drugs they show higher drug loading, stability and biocompatibility, circulation time and localization. Polymeric micelle is an ideal carrier system for both the passive and active target- ed drug delivery as they improve the therapeutic window and reduce the toxic side effects. By using polymeric na- nocarriers the Active Pharmaceutical Ingredients can be protected against biodegradation in the blood circulation and consequently the circulation and the action can be prolonged. The drug molecules are directly and selec- tively delivered to the targeted area by using both active and passive delivery methods. Polymeric micelles show specific strength in solubilizing hydrophobic drugs and reduce the limitations associated with the toxic solvents [78-79].

Crielaard et al., 2012 developed novel derivatives of dex- amethasone. These derivatives are covalently entrapped in polymeric micelles. They are hydrolytically cleavable and specially designed to achieve effective glucocorticoid targeting. The release rate of dexamethasone tried to be controlled by varying the degree of oxidation the thioether in the drug-linker [80].

Sethi et al., 2013 prepared sterically stabilized micelles (SSM) containing low doses of the vasoactive intestinal peptide (VIP) for the treatment of RA. The interaction of SSM and VIP protected the peptide from degradation and prolonged its retention time. The low doses of VIP in SSM can be vasoactive intestinal peptide a novel nanomedi- cine for RA. It was found that it can down-regulate both autoimmune and inflammatory components of RA [81].

4.4. Microemulsion / Nanoemulsion

Nanoemulsions are also known as mini-emulsion or submicron emulsions. It basically consists of oil in water type of emulsion. It possesses the droplet size of 100 - 500 nm. They are the good transporter for lipophilic com- pounds as they possess lipophilic core. They show good stability and clarity during storage. The property of pos- sessing a low viscosity of nanoemulsions is beneficial for preparing sprays [82]. The small size of nanoemulsions provides properties such as high surface area per unit vol-

ume, stability, transparency, and tunable rheology. Gen- erally, two methods are used to prepare nanoemulsions i.e. high energy and low energy method. Other methods include high-pressure homogenization, phase inversion, ultrasonication, bubble bursting method, etc.

Nanoemulsions come under ultrafine dispersion sys- tems and their properties including visual, visco-elastic and targeting properties make them highly efficient nov- el delivery systems. Singh et al., 2017 & Pey et al., 2006, studied and optimized the composition and preparation method of nanoemulsions. They used a factorial design for the study. They concluded that the size of the drop- lets and the polydispersity affected by the variation in the composition and preparation methods. They have also used a central composite design for optimization [83-84].

Shakeel et al., 2008 examined the anti-inflammatory effect of celecoxib nanoemulsions. They compared the prepared transdermal preparation of celecoxib with conventional celecoxib gel on carrageenan-induced paw edema. They used ANOVA for the study. They found that the inflamma- tory response can be enhanced by using nanoemulsions.

They gave a conclusion that nanoemulsions can be successfully used for enhancing the anti-inflammato- ry effects of celecoxib [85]. Mello S B, et al., 2016 tested the anti-inflammatory efficacy of lipid nanoemulsions containing methotrexate given intravenously in rabbits with antigen-induced arthritis. They compared their for- mulation with marketed methotrexate. HPLC method was used for determining the pharmacokinetics of MTX nanoemulsions. They found the result that the uptake of methotrexate nanoemulsion was increased by two folds in arthritic joints. They concluded that the intravenously ad- ministered nanoemulsion was superior to the marketed preparation of MTX [86]. Modi et al., 2011 investigated the efficiency of aceclofenac nanoemulsion applied topical- ly. They used the spontaneous emulsification method for the preparation of oil in water nanoemulsions. They used rat abdominal skin and determined topical permeation by Franz diffusion cell. They compared nanoemulsion gel with a conventional gel of aceclofenac. The result showed an increase in permeability parameters such as perme- ability coefficient, steady-state flux, etc. They concluded that nanoemulsion is potential delivery vehicles for ace- clofenac when applied transdermally [87].

4.5. Nanogels

Nanogels are the polymeric delivery systems having size up to about 500 nm. Generally, nanoparticles are pre- pared and incorporated on the gel base. These systems possess the potential for targeting different types of drugs due to their small particle size, high biocompatibility, bi- odegradability high drug loading, good permeation capa- bilities, high water retention, etc [88]. Drugs from the na- nogels can be delivered passively or actively. Nanogels are basically hydrogel that can accommodate a large amount of water inside them and swell to a large volume thus in- creasing the capacity to load a large amount of drug. Chen et al., 2013, Samah et al., 2010 prepared nanogel of metho- trexate by surfactant-free emulsion polymerization meth-

(12)

od and tested its permeation efficiency by Franz diffusion cells using a porcine ear skin. The presence of nanopar- ticles in the receptors was presented by the TEM study.

They concluded that the enhanced permeation of the drug occurs due to the permeation efficiency of the na- nogels [89-90]. Nagai, N., et al., 2015 prepared and evalu- ated the indomethacin nanogel ointment in adjuvant-in- duced arthritic rats. The indomethacin gel ointment was prepared by bead smash 12. The size of the nanoparticle of indomethacin was about 173 nm. They found the result that the increase in hind paw edema is reduced and the drug concentration in the affected tissues is increased by using nanogel ointment of the drug.

They suggested by their findings that the use of nano- particles, especially when applied topically avoids the unwanted side effects of any drug with a variety of ad- vantages [91]. Khurana et al. 2013 formulated and eval- uated the potential of meloxicam SLN incorporated in a gel base. The formulation was optimized by using various parameters. The skin permeation was determined using Franz diffusion cells and skin tolerance was determined in vivo by histopathological examination using mice. Car- rageenan-induced rat paw edema method was used for determining the anti-inflammatory potential of the gel containing SLN.

The result showed that nano-sized meloxicam SLN pos- sesses controlled-release abilities with good permeation capacity. They gave a concluding remark that SLN gel can be used as an efficient delivery system for meloxicam for the alleviation of inflammatory diseases [92]. Elkomy et al., 2018 investigated the potential of SLN gel of tenoxicam and performed a pharmacokinetic Pharmacodynamic model for determining concentration-time profile in the skin. They used 23 factorial design to study the effect of different formulation variables on the properties of SLN gel for optimization. Gel tolerance was determined by us- ing rabbit skin irritation test and the anti-inflammatory effect was determined by the rat paw edema model. They concluded that SLN- gel is a promising delivery vehicle for tenoxicam through the skin for arthritic disorder and PK- PD modeling is an efficient approach for indirect quanti- tation of skin accumulation [93].

5. CONCLUSION

The pathophysiology behind rheumatoid arthritis (RA) is a complex milieu that, could not be fully described till date. However, the scientific workflow has explained several breakthroughs involved in the development of the disease. Despite this enormous advancement in the methods of treatment of the disease, there is still a lot of scope for further research as there is no therapy available which can completely cure the disease. Presently, maxi- mum therapies work on symptoms of the disease. Along with this, they show poor efficacy, severe side effects, high doses, greater frequency of administration and high cost.

The novel drug delivery systems overcome many of the problems associated with conventional dosage forms.

Passive and active targeting methods deliver the drugs di- rectly to the inflamed site. In spite of these novel efforts,

patients do not respond at all to the therapy in many cases.

At present, a large number of researches of different drugs are reported to the management of rheumatoid arthritis, but a limited number of formulations entered into clinical trials. The field still requires prodigious study.

CONFLICT OF INTEREST

No conflict of interest

Reference

Klareskog L, Padyukov L, Lorentzen J, Alfredsson, L.

Mechanisms of disease: genetic susceptibility and en- vironmental triggers in the development of rheumatoid arthritis. Nat Clin Pract Rheumatol. 2006;2(8):425–33.

Lawrence T, Natoli G. Transcriptional regulation of Macrophage polarization: Enabling diversity with identity. Nat Rev Immunol. 2011;11(11):750–61.

Firestein GS. Evolving Concepts of Rheumatoid Arthri- tis. Nature. 2003;423 (6937):356–61.

Ferrari M, Onuoha SC, Pitzalis C. Going with the flow:

harnessing the power of the vasculature for targeted therapy in rheumatoid arthritis. Drug Discovery Today.

2016;21(1):172–9.

Wright HL, Moots RJ, Edwards SW. The multifactori- al role of neutrophils in rheumatoid arthritis. Nat Rev Rheumatol. 2014;10:593–601.

Koenders MI, Vandenberg WB. Novel therapeutic tar- gets in rheumatoid arthritis. Trends Pharmacol Sci.

2015;36:189–95.

Helmick CG, Felson DT, Lawrence RC, Gabriel S, Hirsch R, Kwoh CK. Estimates of the prevalence of ar- thritis and other rheumatic conditions in the United States: part I. Arthritis Rheum. 2008;58:15–25.

William DA, Lemke TE. Non-steroidal anti-inflamma- tory drug. Foye's Medicinal Chemistry. 5th ed. Phila- delphia: Lippincott, Williams and Wilkins. 2006;751-93.

Clària J. Cyclooxygenase-2 Biology. Current Pharma- ceutical Design. 2003; 9(27):2177-90.

Ribbens C, Andre B, Kaye O, Kaiser MJ, Bonnet V, de Groote D. Increased synovial fluid levels of interleu- kin-12, sCD25, and sTNF-RII/sTNF-RI ratio delineate a cytokine pattern characteristic of immune arthropa- thies. Eur Cytokine Netw. 2000;11(4): 669-76.

Yang L, Zhang J, Wang G. The effect of sodium hyalu- ronate treating knee osteoarthritis on synovial fluid in- terleukin-1β and clinical treat¬ment mechanism. Pak J Pharm Sci. 2015;28(1):407–10.

Moura CC, Segundo MA, Neves J, Reis S, Sarmento B.

Co-association of methotrexate and SPIONs into an- ti-CD64 antibody-conjugated PLGA nanoparticles for theranostic application. Int J Nanomed. 2014;9:4911–

22.

Mashaghi S, Jadidi T, Koenderink G, Mashaghi A. Lipid nanotechnology. Int J Mol Sci 2013;14: 4242–82.

Sehgal R. Nanotechnology and its applications in drug delivery. A review. Int J Mol Med. 2013;3:267.

1.

2.

3.

4.

5.

6.

7.

8.

9.

10.

11.

12.

13.

14.

참조

관련 문서

Tenders are invited for the Supply and Delivery of Vehicles/Mechanical Equipment to the Princes Town Regional Corporation, Ministry of Rural Development and

The side effects of the currently used drug made us to explore an alternative and traditional approach to finding out new drug compound from the natural Flavonoid,

In this study, we found that it was difficult to achieve both high encapsulation efficiency and sustained drug release for drug compound, SS-AG20, using the oil-in-water

But further skin prick test with ranitidine (50 mg/mL), almagate, alibendol, and levosulpiride as it is revealed strong positive reaction to ranitidine only (Fig. 1), and the

Description Drug core surrounded by polymer membrane which controls release rate.. Advantages Zero-order

Hagen, “The Efficacy Requirements of the Food, Drug and Cosmetic Act”, Food Drug Cosm.. Bodenheimer, “The drug Amendments of 1962 : The anatomy of a Regulatory

Basic aspects of AUTOSAR architecture and methodology Safety mechanisms supported by AUTOSAR.. Technical safety concepts supported by AUTOSAR Relationship to ISO

1 John Owen, Justification by Faith Alone, in The Works of John Owen, ed. John Bolt, trans. Scott Clark, &#34;Do This and Live: Christ's Active Obedience as the